EP1463942B1 - Proteinreinigungsverfahren - Google Patents

Proteinreinigungsverfahren Download PDF

Info

Publication number
EP1463942B1
EP1463942B1 EP02794380A EP02794380A EP1463942B1 EP 1463942 B1 EP1463942 B1 EP 1463942B1 EP 02794380 A EP02794380 A EP 02794380A EP 02794380 A EP02794380 A EP 02794380A EP 1463942 B1 EP1463942 B1 EP 1463942B1
Authority
EP
European Patent Office
Prior art keywords
protein
recombinant fusion
chromatography
antibody
tnfr
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Lifetime
Application number
EP02794380A
Other languages
English (en)
French (fr)
Other versions
EP1463942A4 (de
EP1463942A2 (de
EP1463942B2 (de
Inventor
Ganesh Vedantham
Clayton A. Brooks, Iii
Joanne M. Reeder
Andrew M. Goetze
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Immunex Corp
Original Assignee
Immunex Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=27407561&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=EP1463942(B1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Immunex Corp filed Critical Immunex Corp
Publication of EP1463942A2 publication Critical patent/EP1463942A2/de
Publication of EP1463942A4 publication Critical patent/EP1463942A4/de
Publication of EP1463942B1 publication Critical patent/EP1463942B1/de
Application granted granted Critical
Publication of EP1463942B2 publication Critical patent/EP1463942B2/de
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/715Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
    • C07K14/7151Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons for tumor necrosis factor [TNF], for lymphotoxin [LT]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • C07K1/16Extraction; Separation; Purification by chromatography
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • C07K1/16Extraction; Separation; Purification by chromatography
    • C07K1/22Affinity chromatography or related techniques based upon selective absorption processes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70575NGF/TNF-superfamily, e.g. CD70, CD95L, CD153, CD154
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto

Definitions

  • the invention relates to protein purification, in particular, purification utilizing hydroxyapatite chromatography.
  • Proteins A and G are often employed to purify antibodies by affinity chromatography. See e . g . R. Vola et al. (1994), Cell Biophys. 24-25: 27-36 ; Aybay and Imir (2000), J. Immunol. Methods 233(1-2): 77-81 ; Ford et al. (2001), J. Chromatogr. B 754: 427-435 ; DE 42 05 938 . These proteins are useful because they bind to a constant (F C ) portion of many different antibodies. Recombinant fusion proteins including an F C portion of an IgG antibody can be purified using similar methods.
  • Protein A is immunogenic and, in large amounts, potentially toxic. Some Protein A can leach into a sample during affinity chromatography when Protein A affixed to a solid support is used as an adsorbent. See e . g . Bensinger et al. (1984), J. Biol. Response Modif. 3: 347-351 ; Ventura et al. (1987), Cancer Treat. Rep. 71(4): 411-413 .
  • Bispecific monoclonal antibodies have been separated from other antibodies by binding and eluting them from hydroxyapatite subsequent to a pre-purification by affinity chromatography using Protein A affixed to a solid support as an adsorbent. Tarditi et al. (1992), J. Chromatography 599: 13-20 ; Ford et al. (2001), J. Chromatography B 754: 427-435 . Monoclonal antibodies have also been bound and eluted from hydroxyapatite. Ahmad (1999), Ceramic Hydroxyapatite as a Polishing Step for Monoclonal Antibody Purification, Abstract from the Recovery of Biological Products Meeting #9 at Whistler, British Columbia.
  • the present invention provides a simplified and broadly applicable process for using hydroxyapatite chromatography for purification of antibodies and other proteins.
  • Affinity chromatography is a powerful tool for purification of proteins such as antibodies and F C -fusion proteins.
  • proteins are manufactured for therapeutic use, the presence of other proteins, including a protein used as part of an affinity adsorbent, which can leach into a sample during affinity chromatography, is of concern.
  • other protein contaminants may also be present in a sample, such as, for example, proteins derived from host cells that produce the protein being purified.
  • the invention provides, among other things, a high yield technique of solving these problems through hydroxyapetite chromatography in a flow-through mode which involves minimal processing.
  • the invention provides, in one aspect, a method for separating a protein from a second protein comprising subjecting the protein to hydroxyapatite chromatography when (1) the protein has been previously purified by affinity chromatography using the second protein affixed to a solid support as an adsorbent, (2) the second protein can bind to a constant antibody immunoglobulin domain, and (3) the protein does not bind to hydroxyapatite while the second protein does bind to hydroxyapatite under the conditions used.
  • the protein may comprise an F C portion of an antibody.
  • the protein may be TNFR:F C .
  • the second protein may be Protein A or Protein G.
  • the invention further provides a method for purifying a protein from a sample comprising the protein and at least one protein contaminant comprising subjecting the sample to hydroxyapatite chromatography, wherein the protein is separated from at least one protein contaminant by hydroxyapatite chromatography in a solution in which hydroxyapatite chromatography is performed, wherein the majority of molecules of the protein are recovered in the flow through and wash, wherein the protein has been previously purified by affinity chromatography using a second protein affixed to a solid support as an adsorbent, and wherein the second protein binds to a constant antibody immunoglobulin domain.
  • the second protein may or may not be present at a detectable level in the sample, and the protein can be separated from the second protein by hydroxyapatite chromatography in the solution used.
  • the protein and at least one protein contaminant may have been secreted into a culture medium by cultured animal cells, such as CHO cells.
  • the protein may comprise an F C portion of an antibody and may, for example, be TNFR:F C or an antibody.
  • the second protein may be Protein A and Protein G.
  • the solution in which hydroxyapatite chromatography occurs can comprise a sodium phosphate buffer at a concentration between about 5 millimolar and about 50 millimolar, optionally between about 15 millimolar and about 35 millimolar, and can have a pH between about 6.0 and about 8.6.
  • the invention further provides, in another aspect, a method for separating a recombinant fusion protein from a second protein comprising subjecting the recombinant fusion protein to hydroxyapatite chromatography when (1) the recombinant fusion protein has been previously purified by affinity chromatography using the second protein affixed to a solid support as an adsorbent and (2) the recombinant fusion protein comprises an F C portion of an antibody and part or all of a non-antibody protein.
  • the recombinant fusion protein can be TNFR:F C .
  • the invention further encompasses a method for separating a recombinant fusion protein from a second protein comprising subjecting the recombinant fusion protein to hydroxyapatite chromatography when (1) the recombinant fusion protein does not bind to hydroxyapatite and the second protein does bind to hydroxyapatite under the conditions used, (2) the recombinant fusion protein has been previously purified by affinity chromatography using the second protein affixed to a solid support as an adsorbent, and (3) the recombinant fusion protein comprises the F C region of an antibody.
  • the invention provides a method for purifying a recombinant fusion protein from sample comprising the recombinant fusion protein and at least one protein contaminant comprising subjecting the sample to hydroxyapatite chromatography, wherein the recombinant fusion protein comprises an F C portion of an antibody and part or all of a non-antibody protein, and wherein the recombinant fusion protein has been previously purified by affinity chromatography using a second protein affixed to a solid support as an adsorbent.
  • the majority of molecules of the recombinant fusion protein can be recovered in the flow through and wash.
  • the recombinant fusion protein can be TNFR:F C
  • the second protein can be Protein A or Protein G.
  • the hydroxyapatite chromatography can be carried out in a solution comprising a sodium phosphate buffer, optionally at a concentration between 5 millimolar and about 50 millimolar or between about 15 millimolar and about 35 millimolar and optionally at a pH between about 6.0 and about 8.6.
  • the recombinant fusion protein and/or at least one protein contaminant can be been secreted into a culture medium by cultured animal cells, optionally CHO cells.
  • a method for separating TNFR:F C from Protein A comprising subjecting TNFR:F C to hydroxyapatite chromatography subsequent to purifying TNFR:F C by affinity chromatography using Protein A affixed to a solid support as an adsorbent.
  • the invention provides a method for purifying TNFR:F C comprising subjecting a sample comprising TNFR:F C and at least one protein contaminant to hydroxyapatite chromatography under conditions wherein the majority of molecules of TNFR:F C are recovered in the flow through and wash.
  • a further embodiment provides a method for purifying TNFR:F C comprising subjecting TNFR:F C to hydroxyapatite chromatography under conditions where TNFR:F C does not bind to hydroxyapatite, whereby TNFR:F C is separated from at least one protein contaminant.
  • the invention further provides, in still another aspect, a method for separating a recombinant fusion protein from host cell proteins comprising loading a sample comprising the recombinant fusion protein onto hydroxyapatite, subjecting the recombinant fusion protein to hydroxyapatite chromatography under conditions where the recombinant fusion protein does not bind to hydroxyapatite, and recovering a sample comprising the recombinant fusion protein in the combined flow through and wash.
  • a method for separating a protein from host cell proteins comprising subjecting a sample comprising the protein and at least one host cell protein to hydroxyapatite chromatography, whereby the protein is separated from at least one host cell protein, wherein the protein comprises a constant antibody immunoglobulin domain, wherein the majority of molecules of the protein are recovered in the flow through and wash, and wherein both the protein and the host cell protein have been secreted into a culture medium by cultured animal cells, such as, for example CHO cells.
  • the concentration of the host cell proteins can be less than about 100 parts per million, and the.protein can be TNFR:F C .
  • TNFR:F C TNFR:F C
  • the preparation comprises less than 3 parts per million Protein A and less than 100 parts per million host animal cell proteins, optionally less than 2 parts per million Protein A and/or less than 75 parts per million host cell proteins.
  • Figure 1 shows the elution profile of a hydroxyapatite column run in 25 millimolar sodium phosphate, pH 6.8 onto which a sample comprising TNFR: F C as a majority species was loaded, as described in Example 2.
  • Adsorbent is at least one molecule affixed to a solid support or at least one molecule that is, itself, a solid, which is used to perform chromatography.
  • Affinity chromatography is chromatography that utilizes the specific, reversible interactions between biomolecules, for example, the ability of Protein A to bind to an F C portion of an IgG antibody, rather than the general properties of a molecule, such as isoelectric point, hydrophobicity, or size, to effect chromatographic separation.
  • affinity chromatography involves using an adsorbent, such as Protein A affixed to a solid support, to chromatographically separate molecules that bind more or less tightly to the adsorbent. See Ostrove (1990) in Guide to Protein Purification, Methods in Enzymology 182: 357-379 .
  • Antibody is a protein or complex of proteins, each of which comprises at least one variable antibody immunoglobulin domain and at least one constant antibody immunoglobulin domain.
  • Antibodies may be single chain antibodies, dimeric antibodies, or some higher order complex of proteins including, but not limited to, heterodimeric antibodies.
  • Chromatography is the separation of chemically different molecules in a mixture from one another by percolation of the mixture through an adsorbent, which absorbs or retains different molecules more or less strongly. Molecules that are least strongly adsorbed to or retained by the adsorbent are released from the adsorbent under conditions where those more strongly adsorbed or retained are not.
  • Constant antibody immunoglobulin domain is an immunoglobulin domain that is identical to or substantially similar to a C L , C H 1, C H 2, C H 3, or C H 4, domain of human or animal origin. See e . g . Charles A Hasemann and J. Donald Capra, Immunoglobulins: Structure and Function, in William E. Paul, ed., Fundamental Immunology, Second Edition, 209, 210-218 (1989 ).
  • a contaminant is any foreign or objectable molecule, particularly a biological macromolecule such as a DNA, an RNA, or a protein, other than the protein being purified that is present in a sample of a protein being purified.
  • Contaminants include, for example, other proteins from cells that secrete the protein being purified and proteins, such as Protein A, that are part of an adsorbent used for affinity chromatography that may leach into a sample during affinity chromatography.
  • the F C portion of an antibody includes human or animal immunoglobulin domains C H 2 and C H 3 or immunoglobulin domains substantially similar to these.
  • the biological activity of an F C portion of an antibody for the purpose of determining substantial similarity is the ability to be bound by a second protein that binds to naturally-occurring F C portions of antibodies, such as Protein A or Protein G.
  • a second protein that binds to naturally-occurring F C portions of antibodies, such as Protein A or Protein G.
  • Host cell proteins are proteins encoded by the naturally-occurring genome of a host cell into which DNA encoding a protein that is to be purified is introduced. Host cell proteins may be contaminants of the protein to be purified, the levels of which may be reduced by purification. Host cell proteins can be assayed for by any appropriate method including gel electrophoresis and staining and/or ELISA assay, among others.
  • Hydroxyapatite chromatography is chromatography using ceramic hydroxyapatite as an adsorbent. See e.g. Marina J. Gorbunoff (1990), Protein Chromatography on Hydroxyapatite Columns, in Guide to Protein Purification, Murray P. Deutscher, ed., Methods in Enzymology 182: 329-339 .
  • an IgG antibody is an antibody that includes at least one ⁇ type constant immunoglobulin domain. For discussion, see Hasemann and Capra, supra , at 226.
  • Polypeptide For the purposes of the invention, “polypeptide” is used interchangeably with “protein.”
  • a protein is any chain of at least five amino acids linked by peptide bonds.
  • Protein A is a protein originally discovered in the cell wall of Stapphylococcus that binds specifically to an F C portion of IgG antibody.
  • Protein A is any protein identical or substantially similar to Stapphylococcal Protein A, including commercially available and/or recombinant forms of Protein A.
  • the biological activity of Protein A for the purpose of determining substantial similarity is the capacity to bind to an F C portion of IgG antibody.
  • Protein G is a protein originally discovered in the cell wall of Streptococcus that binds specifically to an F C portion of an IgG antibody.
  • Protein G is any protein identical or substantially similar to Streptococcal Protein G, including commercially available and/or recombinant forms of Protein G.
  • the biological activity of Protein G for the purpose of determining substantial similarity is the capacity to bind to an F C portion of IgG antibody.
  • Protein LG is a recombinant fusion protein that binds to IgG antibodies comprising portions of both Protein G (see definition above) and Protein L. Protein L was originally isolated from the cell wall of Peptostreptococcus. Protein LG comprises IgG binding domains from both Protein L and G. Vola et al. (1994) Cell. Biophys. 24-25: 27-36 .
  • Protein LG is any protein identical or substantially similar to Protein LG, including commercially available and/or recombinant forms of Protein LG.
  • the biological activity of Protein LG for the purpose of determining substantial similarity is the capacity to bind to an IgG antibody.
  • To purify a protein means to reduce the amounts of foreign or objectionable elements, especially biological macromolecules such as proteins or DNA, that may be present in a sample of the protein.
  • the presence of foreign proteins may be assayed by any appropriate method including gel electrophoresis and staining and/or ELISA assay.
  • the presence of DNA may be assayed by any appropriate method including gel electrophoresis and staining and/or assays employing polymerase chain reaction.
  • a recombinant fusion protein is any protein that comprises part or all of two or more proteins that are not fused in their natural state. Examples of such proteins include, but are not limited to, human receptor activator of NF-KappaB fused to an F C portion of an antibody (huRANK: F C ), tunica internal endothelial cell kinase-delta fused to an F C portion of an antibody (TEKdelta: F C ), and tumor necrosis factor receptor fused to an F C portion of an antibody (TNFR: F C ).
  • a protein is separated from a second protein in a mixture comprising both proteins when the mixture is subjected to a process such that at least the majority of the molecules of the protein are removed from that portion of the mixture that comprises at least the majority of the molecules of the second protein.
  • proteins are substantially similar if they are at least 80%, preferably at least 90% identical to each other in amino acid sequence and maintain or alter in a desirable manner the biological activity of the unaltered protein. Included in amino acids considered identical for the purpose of determining whether proteins are substantially similar are amino acids that are conservative substitutions, unlikely to affect biological activity, including the following: Ala for Ser, Val for Ile, Asp for Glu, Thr for Ser, Ala for Gly, Ala for Thr, Ser for Asn, Ala for Val, Ser for Gly, Tyr for Phe, Ala for Pro, Lys for Arg, Asp for Asn, Leu for Ile, Leu for Val, Ala for Glu, Asp for Gly, and these changes in the reverse.
  • the percent identity of two amino sequences can be determined by visual inspection and mathematical calculation, or more preferably, the comparison is done by comparing sequence information using a computer program such as the Genetics Computer Group (GCG; Madison, WI) Wisconsin package version 10.0 program, 'GAP' ( Devereux et al., 1984, Nucl. Acids Res. 12: 387 ) or other comparable computer programs.
  • GCG Genetics Computer Group
  • 'GAP' Devereux et al., 1984, Nucl. Acids Res. 12: 387
  • the preferred default parameters for the 'GAP' program includes: (1) the weighted amino acid comparison matrix of Gribskov and Burgess ((1986), Nucl. Acids Res.
  • TNFR refers to proteins comprising amino acid sequences that are identical or substantially similar to the sequence of a native mammalian tumor necrosis factor receptor (TNFR).
  • Biological activity for the purpose of determining substantial similarity means the capacity to bind tumor necrosis factor (TNF), to transduce a biological signal initiated by TNF binding to a cell, or to cross-react with anti-TNFR antibodies raised against TNFR from natural (i.e., non-recombinant) sources.
  • a TNFR may be any mammalian TNFR, including murine or human TNFRs. Such TNFRs are described in U.S. Patent No. 5,395,760 , and in U.S. Patent No. 5,610,279 .
  • a particularly preferred TNFR is that described in US Patent No. 5,395,760 , which has an apparent molecular weight by SDS-PAGE of about 80 kilodaltons in its glycosylated form.
  • TNFR F C is a recombinant fusion protein comprising all or part of an extracellular domain of a TNFR fused to an F C region of an antibody.
  • Such an extracellular domain includes, but is not limited to, amino acid sequences substantially similar to amino acids 1-163, 1-185, or 1-235 of Figure 2A of US Patent No. 5,395,760 .
  • Possible uses of TNFR:Fc purified by conventional methods are discussed in Srivastava (2001) Research Communications in Molecular Pathology and Pharmacology 109(1 & 2):125-141 ; Strand (1999) Clinical Cornerstones Office Rheumatology 2(2):38-50 ; Hodge et al (1998) Immunology Letters 63:49-51 .
  • variable antibody immunoglobulin domain is an immunoglobulin domain that is identical or substantially similar to a V L or a V H domain of human or animal origin.
  • the biological activity of a variable antibody immunoglobulin domain for the purpose of determining substantial similarity is antigen binding.
  • the process of purifying a protein often involves numerous steps.
  • the present invention encompasses a process for reducing the amount of a second protein in a mixture comprising a protein that is being purified and a second protein, wherein the second protein is introduced during an affinity chromatography step in which the second protein is part of the adsorbent. Removal of such a second protein can be challenging when the isoelectric points of the protein being purified and a complex of the second protein with the protein being purified are close because ion exchange chromatography is unlikely to effect a separation of such proteins. Use of hydroxyapatite makes separation both possible and simple.
  • the methods of the invention also have the added advantage of removing other objectionable matter from the protein.
  • the invention comprises a method for purifying a protein comprising an F C region of an antibody using hydroxyapatite chromatography under conditions where the protein is recovered in the flow through and wash and at least one protein contaminant is retained on the hydroxyapatite.
  • the methods of the invention can reduce the amount of a second protein, and/or a complex of the second protein with the protein being purified, which is introduced during affinity chromatography, in a sample that contains the protein being purified.
  • hydroxyapatite chromatography is performed under conditions such that the protein being purified does not bind to hydroxyapatite, but the second protein, and/or a complex of the second protein with the protein being purified, does bind.
  • the process of the invention can, in some embodiments, also involve at least two steps.
  • the entire process of purifying the protein may include other steps before and/or after each of these steps.
  • the hydroxyapatite chromatography medium may be pre-equilibrated in a chosen solution, e.g. a salt and/or buffer solution.
  • Pre-equilibration serves the function of displacing a solution used for regenerating and/or storing the chromatography medium.
  • a chosen solution e.g. a salt and/or buffer solution.
  • Pre-equilibration serves the function of displacing a solution used for regenerating and/or storing the chromatography medium.
  • appropriate pre-equilibration solutions may include the same buffer or salt used for performing the chromatography, optionally, at a higher concentration than is used to perform chromatography. Buffers and salts that can be used for chromatography are discussed below.
  • pre-equilibration may take place in a in a solution comprising sodium phosphate at a higher concentration.
  • pre-equilibration may occur in a solution comprising sodium phosphate at concentrations between about 0.2 molar and about 0.5 molar, more preferably in concentrations of sodium phosphate between about 0.3 molar and about 0.4 molar, inclusive.
  • the hydroxyapatite chromatography medium can be equilibrated in the buffer or salt that will be used to chromatograph the protein.
  • chromatography and loading of the protein to be purified
  • buffers or salts including sodium, potassium, ammonium, magnesium, calcium, chloride, fluoride, acetate, phosphate, and/or citrate salts and/or Tris buffer.
  • buffers or salts can have a pH of at least about 5.5.
  • equilibration may take place in a solution comprising a Tris or a sodium phosphate buffer.
  • the sodium phosphate buffer is at a concentration between about 0.5 millimolar and about 50 millimolar, more preferably at a concentration between about 15 millimolar and 35 millimolar.
  • equilibration takes place at a pH of at least about 5.5.
  • Equilibration may take place at pHs between about 6.0 and about 8.6, preferably at pHs between about 6.5 and 7.5.
  • the solution comprises a sodium phosphate buffer at a concentration of about 25 millimolar and at a pH of about 6.8.
  • Chromatography may be carried out in a column.
  • the column may be run with or without pressure and from top to bottom or bottom to top. The direction of the flow of fluid in the column may be reversed during the chromatography process.
  • Chromatography may also be carried out using a batch process in which the solid support is separated from the liquid used to load, wash, and elute the sample by any suitable means, including gravity, centrifugation, or filtration.
  • Chromatography may also be carried out by contacting the sample with a filter that adsorbs or retains some molecules in the sample more strongly than others.
  • the protein can be produced by living host cells that have been genetically engineered to produce the protein.
  • Methods of genetically engineering cells to produce proteins are well known in the art. See e.g. Ausabel et al., eds. (1990), Current Protocols in Molecular Biology (Wiley, New York ). Such methods include introducing nucleic acids that encode and allow expression of the protein into living host cells.
  • These host cells can be bacterial cells, fungal cells, or, preferably, animal cells grown in culture.
  • Bacterial host cells include, but are not limited to, Escherichia coli cells. Examples of suitable E. coli strains include: HB101, DH5 ⁇ , GM2929, JM109, KW251, NM538, NM539, and any E.
  • Fungal host cells that can be used include, but are not limited to, Saccharomyces cerevisiae, Pichia pastoris and Aspergillus cells.
  • a few examples of animal cell lines that can be used are CHO, VERO, BHK, HeLa, Cos, MDCK, 293, 3T3, and WI38. New animal cell lines can be established using methods well know by those skilled in the art (e.g., by transformation, viral infection, and/or selection).
  • the protein can be secreted by the host cells into the medium.
  • Protein concentration of a sample at any stage of purification can be determine by any suitable method. Such methods are well known in the art and include: 1) colorimetric methods such as the Lowry assay, the Bradford assay, the Smith assay, and the colloidal gold assay; 2) methods utilizing the UV absorption properties of proteins; and 3) visual estimation based on stained protein bands on gels relying on comparison with protein standards of known quantity on the same gel. See e.g. Stoschek (1990), Quantitation of Protein, in Guide to Protein Purification, Methods in Enzymol. 182: 50-68 .
  • the protein undergoing purification as contemplated by the invention comprises one or more constant antibody immunoglobulin domain(s) and may, but need not, comprise a single or multiple variable antibody immunoglobulin domain(s). It may be a naturally-occurring protein or a recombinant fusion protein. It may comprise an F C portion of an antibody. It may also comprise a non-antibody protein.
  • proteins specifically contemplated for use with the invention include recombinant fusion proteins comprising one or more constant antibody immunoglobulin domains, optionally an F C portion of an antibody, and a protein identical to or substantially similar to one of the following proteins: a flt3 ligand (as described in international application no. WO 94/28391 ), a CD40 ligand (as described in US Patent No.
  • erythropoeitin thrombopoeitin, calcitonin
  • Fas ligand ligand for receptor activator of NF-kappa B (RANKL), tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL, as described in international application no. WO 97/01633 ), thymic stroma-derived lymphopoietin, granulocyte colony stimulating factor, granulocyte-macrophage colony stimulating factor (GM-CSF, as described in Australian Patent No.
  • Proteins contemplated by the invention also include recombinant fusion proteins comprising one or more constant antibody immunoglobulin domains, optionally an F C portion of an antibody, plus a receptor for any of the above-mentioned proteins or proteins substantially similar to such receptors.
  • These receptors include: both forms of TNFR (referred to as p55 and p75), Interleukin-1 receptors types I and II (as described in EP Patent No. 0 460 846 , US Patent No. 4,968,607 , and US Patent No. 5,767,064 ), Interleukin-2 receptor, Interleukin-4 receptor (as described in EP Patent No. 0 367 566 and US Patent No.
  • Interleukin-15 receptor Interleukin-17 receptor, Interleukin-18 receptor, granulocyte-macrophage colony stimulating factor receptor, granulocyte colony stimulating factor receptor, receptors for oncostatin-M and leukemia inhibitory factor, receptor activator of NF-kappa B (RANK, as described in US Patent No. 6,271,349 ), receptors for TRAIL (including TRAIL receptors 1, 2, 3, and 4), and receptors that comprise death domains, such as Fas or Apoptosis-Inducing Receptor (AIR).
  • TRAIL including TRAIL receptors 1, 2, 3, and 4
  • AIR Apoptosis-Inducing Receptor
  • CD proteins differentiation antigens
  • ligands or proteins substantially similar to either of these which are fused to at least one constant antibody immunoglobulin domain, optionally an F C portion of an antibody.
  • antigens are disclosed in Leukocyte Typing VI (Proceedings of the VIth International Workshop and Conference, Kishimoto, Kikutani et al., eds., Kobe, Japan, 1996 ). Similar CD proteins are disclosed in subsequent workshops. Examples of such antigens include CD27, CD30, CD39, CD40, and ligands thereto (CD27 ligand, CD30 ligand, etc.).
  • CD antigens are members of the TNF receptor family, which also includes 41BB ligand and OX40.
  • the ligands are often members of the TNF family, as are 41BB ligand and OX40 ligand. Accordingly, members of the TNF and TNFR families can also be purified using the present invention.
  • Enzymatically active proteins or their ligands can also be purified according to the invention.
  • Examples include recombinant fusion proteins comprising at least one constant antibody immunoglobulin domain plus all or part of one of the following proteins or their ligands or a protein substantially similar to one of these: metalloproteinase-disintegrin family members, various kinases, glucocerebrosidase, superoxide dismutase, tissue plasminogen activator, Factor VIII, Factor IX, apolipoprotein E, apolipoprotein A-I, globins, an IL-2 antagonist, alpha-1 antitrypsin, TNF-alpha Converting Enzyme, ligands for any of the above-mentioned enzymes, and numerous other enzymes and their ligands.
  • the method of the invention may also be used to purify antibodies or portions thereof and chimeric antibodies, i . e . antibodies having human constant antibody immunoglobulin domains coupled to one or more murine variable antibody immunoglobulin domain, or fragments thereof.
  • the method of the invention may also be used to purify conjugates comprising an antibody and a cytotoxic or luminescent substance.
  • Such substances include: maytansine derivatives (such as DM1); enterotoxins (such as a Staphlyococcal enterotoxin); iodine isotopes (such as iodine-125); technium isotopes (such as Tc-99m); cyanine fluorochromes (such as Cy5.5.18); and ribosome-inactivating proteins (such as bouganin, gelonin, or saporin-S6).
  • enterotoxins such as a Staphlyococcal enterotoxin
  • iodine isotopes such as iodine-125
  • technium isotopes such as Tc-99m
  • cyanine fluorochromes such as Cy5.5.18
  • ribosome-inactivating proteins such as bouganin, gelonin, or saporin-S6
  • antibodies or antibody/cytotoxin or antibody/luminophore conjugates contemplated by the invention include those that recognize any one or combination of the above-described proteins and/or the following antigens: CD2, CD3, CD4, CD8, CD11a, CD14, CD18, CD20, CD22, CD23, CD25, CD33, CD40, CD44, CD52, CD80 (B7.1), CD86 (B7.2), CD147, IL-1 ⁇ , IL-1 ⁇ , IL-4, IL-5, IL-8, IL-10, IL-2 receptor, IL-4 receptor, IL-6 receptor, IL-13 receptor, IL-18 receptor subunits, PDGF- ⁇ , VEGF, TGF, TGF- ⁇ 2, TGF- ⁇ 1, EGF receptor, VEGF receptor, C5 complement, IgE, tumor antigen CA125, tumor antigen MUC1, PEM antigen, LCG (which is a gene product that is expressed in association with lung cancer), HER-2, a tumor-associated glycoprotein TAG-72, the SK
  • the invention may also be used to purify anti-idiotypic antibodies, or substantially similar proteins, including but not limited to anti-idiotypic antibodies against: an antibody targeted to the tumor antigen gp72; an antibody against the ganglioside GD3; or an antibody against the ganglioside GD2.
  • an adsorbent in affinity chromatography, can comprise a suitable solid support with a second protein affixed to it. A protein sample containing the protein to be purified can be applied to this absorbent. The adsorbent can be subsequently washed in a solution that does not interfere with the binding of the second protein to the constant antibody immunoglobulin domain of the protein. The protein can thereafter be eluted from the adsorbent with a solution that interferes with the binding of the constant antibody immunoglobulin domain by the second protein.
  • the second protein is any protein that can bind to a constant antibody immunoglobulin domain, which may, but need not, be a recombinant fusion protein.
  • the second protein can be Protein G, Protein LG, or Protein A.
  • the second protein can be affixed to any suitable solid support including: agarose, sepharose, silica, collodion charcoal, sand, and any other suitable material. Such materials are well known in the art. Any suitable method can be used to affix the second protein to the solid support. Methods for affixing proteins to suitable solid supports are well known in the art. See e.g. Ostrove (1990), in Guide to Protein Purification, Methods in Enzymology, 182: 357-371 . Furthermore, such solid supports already having the second protein affixed are commercially available from a number of manufacturers including BioRad, Merck, Amersham Pharmacia Biotech, and Millipore Corporation.
  • a protein sample comprising the protein to be purified plus contaminants is loaded onto the adsorbent, which comprises the second protein affixed to a solid support, in a solution comprising a buffer and/or a salt.
  • Suitable buffers include, but are not limited to, phosphate buffers, Tris buffers, acetate buffers, and/or citrate buffers.
  • Suitable salts include, but are not limited to, sodium chloride, potassium chloride, ammonium chloride, sodium acetate, potassium acetate, ammonium acetate, calcium salts, and/or magnesium salts.
  • the solution may comprise Tris at concentrations between about 5 millimolar and 100 millimolar and sodium chloride at concentrations between about 50 millimolar and 250 millimolar.
  • other buffers and salts can be used.
  • the adsorbent can be washed with more of the same solution.
  • the protein can be eluted using a solution that interferes with the binding of the second protein to the constant antibody immunoglobulin domain.
  • This solution may include a chaotropic agent, such as guanidinium, an agent that can either increase or decrease pH, and/or a salt.
  • This solution may include acetic acid, glycine, or citric acid. Elution may be effected by lowering the pH.
  • the pH can be lowered to about 4.5 or less, typically to between about 3.3 to about 4.0, using a solution comprising citrate or acetate, among other possibilities.
  • the pH can be increased, typically to above about 8.5.
  • Solutions appropriate to effect such elutions may comprise Tris or sodium carbonate, among other possibilities.
  • Other methods of elution are also available. Protocols for such affinity chromatography are well known in the art. See e . g . Miller and Stone (1978), J. Immunol. Methods 24(1-2): 111-125 . Conditions for binding and eluting can be easily optimized by those skilled in the art.
  • the protein is subjected to hydroxyapatite chromatography under conditions in which the protein does not bind to hydroxyapatite but the second protein, and/or a complex of the second protein with the protein, which may be present after affinity chromatography, does.
  • the sample is loaded onto hydroxyapatite and chromatography is performed in a solution comprising a buffer and/or a salt at a pH of greater than about 5.5.
  • chromatography can occur in a solution that is the same as or similar to that in which the protein is loaded onto the chromatography medium. Hydroxyapatite chromatography can occur under conditions where the protein and the second protein bind to each other to form a complex or under conditions where they do not.
  • separation of the protein from the second protein can entail separation of the protein from a complex of the second protein with the protein.
  • separation of the protein from the second protein can entail precisely that. Chromatography and loading can occur in a variety of buffers and/or salts including sodium, potassium, ammonium, magnesium, calcium, chloride, fluoride, acetate, phosphate, citrate and/or Tris buffers.
  • buffers and salts are: Tris, sodium phoshate, potassium phophate, ammonium phosphate, sodium chloride, potassium chloride, ammonium chloride, magnesium chloride, calcium chloride, sodium fluoride, potassium fluoride, ammonium fluoride, calcium fluoride, magnesium fluoride, sodium citrate, potassium citrate, ammonium citrate, magnesium acetate, calcium acetate, sodium acetate, potassium acetate, or ammonium acetate.
  • the pH range is chosen to optimize the chromatography conditions and to retain the desired characteristics of the protein of interest. For most proteins of interest, that may range between about 6.0 and about 8.6, preferably between about 6.5 and about 7.5.
  • the loading/chromatography solution comprises a sodium phosphate buffer at a concentration between about 0.5 millimolar and about 50 millimolar, more preferably at a concentration between about 15 millimolar and 35 millimolar sodium phosphate.
  • the solution comprises a sodium phosphate buffer at a concentration of about 25 millimolar and at a pH of about 6.8.
  • the loading solution comprises Tris at a pH between about 6.0 and about 9.0, preferably between about 6.5 and about 8.0.
  • the flow-through liquid which comprises the protein being purified, is collected.
  • the selected buffer and/or salt at the selected concentration allows the second protein to bind to hydroxyapatite, while the protein being purified does not.
  • One skilled in the art will be guided by the knowledge in the art in determining which buffer or salt is appropriate for the particular protein being purified. See e.g. Gorbunoff (1990), Protein Chromatography on Hydroxyapatite Columns, in Guide to Protein Purification, Methods in Enzymology 182: 329-339 ; Scopes, Protein Purification: Principles and Practice, Third Edition, pp. 173-75, Springer, 1994 .
  • a skilled artisan can easily determine the optimal concentration of the selected buffer or salt to use by, for example, running a gradient of the selected buffer or salt through a hydroxyapatite column to which a sample comprising the protein to be purified and the second protein has been applied. Fractions of the effluent of the column can be collected and analyzed to determine the concentration of buffer or salt at which the protein and the second protein elute. Suitable analyses include, for example, a measurement of electrical conductance with a conductivity meter (to determine the salt concentration in the sample) plus gel electrophoresis or ELISA assay (to determine the identity of the proteins in the sample).
  • the hydroxyapatite can be washed with more of the same solution in which the protein sample was loaded, and this wash solution can also be collected and combined with the flow-through liquid.
  • proteins that may remain bound to the hydroxyapatite may be released by stripping the chromatography medium using a solution comprising the buffer or salt used for chromatography, but at a higher molarity. Then, the column may be regenerated using a solution that will have the effect of releasing most or all proteins from the chromatography medium and reducing or eliminating any microbial contamination that may be present in the chromatography medium.
  • a solution may comprise sodium hydroxide.
  • Other reagents can also be used.
  • the column may be rinsed and stored in a solution that can discourage microbial growth.
  • Such a solution may comprise sodium hydroxide, but other reagents can also be appropriate.
  • the second protein, a complex of the protein and the second protein, and/or other proteins that may be present in a sample of the protein being purified can be monitored by any appropriate means.
  • the technique should be sensitive enough to detect contaminants in the range between about 2 parts per million (ppm) (calculated as nanograms per milligram of the protein being purified) and 500 ppm.
  • enzyme-linked immunosorbent assay a method well known in the art, may be used to detect contamination of the protein by the second protein. See e . g . Reen (1994), Enzyme-Linked Immunosorbent Assay (ELISA), in Basic Protein and Peptide Protocols, Methods Mol. Biol. 32: 461-466 .
  • hydroxyapatite chromatography may not detectably reduce contamination by a second protein, especially if the material loaded onto hydroxyapatite has levels of the second protein that are close to or below detectable levels.
  • hydroxyapatite chromatography may reduce contamination by a second protein at least about twofold, preferably at least about threefold, more preferably at least about fivefold, still more preferably at least about tenfold, even more preferably at least about fifteenfold, most preferably at least about twentyfold.
  • contamination of the protein by the second protein after hydroxyapatite chromatography is not more than about 400 ppm, more preferably not more than about 360 ppm, more preferably not more than about 320 ppm, more preferably not more than about 280 ppm, more preferably not more than about 240 ppm, more preferably not more than about 200 ppm, more preferably not more than about 160 ppm, more preferably not more than about 140 ppm, more preferably not more than about 120 ppm, more preferably not more than about 100 ppm, more preferably not more than about 80 ppm, more preferably not more than about 60 ppm, more preferably not more than about 40 ppm, more preferably not more than about 20 ppm, more preferably not more than about 10 ppm, more preferably not more than about 5 ppm, more preferably not more than about 1 ppm, and most preferably not more than about 0.5 ppm.
  • Contamination by such a second protein can range from undetectable levels to about 5 ppm or from about 5 ppm to about 400 ppm. If a protein is being purified for pharmacological use, one of skill in the art will realize that the preferred level of the second protein can depend on the weekly dose of the protein to be administered per patient, with the aim that the patient will not receive more than a certain amount of a contaminating protein per week. Thus, if the required weekly dose of the protein is decreased, the level of contamination by a second protein may possibly increase.
  • contamination of the protein by such other proteins can be reduced after hydroxyapatite chromatography, preferably by at least about twofold, more preferably by at least about threefold, more preferably by at least about fivefold, more preferably by at least about tenfold, more preferably by at least about twentyfold, more preferably by at least about thirtyfold, more preferably by at least about fortyfold, more preferably by at least about fiftyfold, more preferably by at least about sixtyfold, more preferably by at least about seventyfold, more preferably by at least about 80 fold, more preferably by at least about 90 fold, and most preferably by at least about 100 fold.
  • contamination of the protein by such other proteins after hydroxyapatite chromatography is not more than about 10,000 ppm, preferably not more than about 2500 ppm, more preferably not more than about 400 ppm, more preferably not more than about 360 ppm, more preferably not more than about 320 ppm, more preferably not more than about 280 ppm, more preferably not more than about 240 ppm, more preferably not more than about 200 ppm, more preferably not more than about 160 ppm, more preferably not more than about 140 ppm, more preferably not more than about 120 ppm, more preferably not more than about 100 ppm, more preferably not more than about 80 ppm, more preferably not more than about 60 ppm, more preferably not more than about 40 ppm, more preferably not more than about 30 ppm, more preferably not more than about 20 ppm, more preferably not more than about 10 ppm, and most preferably not more than about 5 ppm.
  • Such contamination can range from undetectable levels to about 10 ppm or from about 10 ppm to about 10,000 ppm. If a protein is being purified for pharmacological use, one of skill in the art will realize that the acceptable level of other protein contaminants can depend on the weekly dose of the protein to be administered per patient, as explained above.
  • the amount of DNA that may be present in a sample of the protein being purified can be determined by any suitable method. For example, one can use an assay utilizing polymerase chain reaction. Optionally, the technique can detect DNA contamination at levels of 10 picograms per milligram of protein and greater. DNA levels can be reduced by hydroxyapatite chromatography, optionally by about twofold, preferably by about fivefold, more preferably by about tenfold, more preferably by about fifteenfold, most preferably by about 20 fold.
  • levels of DNA after hydroxyapatite chromatography are less than about 20 picograms per milligram of protein, preferably less than 15 picograms per milligram of protein, more preferably less than 10 picograms per milligram of protein, most preferably less than 5 picograms per milligram of protein.
  • This experiment demonstrates that hydroxyapatite chromatography can reduce levels of residual protein A in a protein sample comprising TNFR:F C that contains a defined amount of protein A, which can form a complex with TNFR:F C .
  • a column of ceramic hydroxyapatite (Type II, Bio-Rad, 80 ⁇ ) 18 cm in height and 1.6 cm in internal diameter was pre-equilibrated with two column volumes of 0.4 molar sodium phosphate, pH 6.8 and equilibrated with four column volumes of 25 millimolar sodium phosphate, pH 6.8, which has a conductivity of 2.8 milliSiemens (mS).
  • a protein sample (5.11 milligrams/milliliter) in 668 milliliters of 25 mM sodium phosphate, pH 6.8 comprising TNFR:F C and protein A (209 ppm) was loaded onto the column. The amount of protein A in the sample was determined using an ELISA assay.
  • the flow-through liquid, containing TNFR:F C was collected.
  • the column was washed with three column volumes of 25 millimolar sodium phosphate, pH 6.8, and the wash was collected and combined with the flow through.
  • the collected protein in the flow through and wash was sterilized by filtration and stored at 2-8°C until the next purification step was carried out. Most of the TNFR:F C loaded is recovered (97%) in the flow-through liquid plus the wash.
  • the amount of protein A in the flow-through plus the wash was measured at 11 ppm.
  • the column was stripped using three column volumes of 0.4 molar sodium phosphate, pH 6.8, regenerated using two column volumes of 1 molar sodium hydroxide, and rinsed with three column volumes of 0.1 molar sodium hydroxide, 10 millimolar sodium phosphate, and stored in the same solution, in which condition it is ready for reuse.
  • the following experiment demonstrates that the levels of host cell proteins in a sample comprising TNFR:F C can be reduced by hydroxyapatite chromatography.
  • a column of ceramic hydroxyapatite (Type II, Bio-Rad, 80 ⁇ ) 10 cm in height and 1.1 cm in internal diameter was pre-equilibrated with two column volumes of 0.3 molar sodium phosphate, pH 6.8 and equilibrated with three column volumes of 25 millimolar sodium phosphate, pH 6.8, which has a conductivity of 2.8 mS.
  • This sample included TNFR:F C , which comprised the vast majority of the sample, and host cell proteins (545 ppm). The amount of host cell proteins in the sample was determined using ELISA assays.
  • the flow-through liquid comprising TNFR:F C
  • the column was washed with three column volumes of 25 millimolar sodium phosphate, pH 6.8, and the wash was collected and combined with the flow through. Most of the TNFR:F C loaded (92%) was recovered in the flow-through liquid plus the wash.
  • the collected TNFR:F C was sterilized by filtration. The amount of host cell proteins was lowered at least about seventy fold (from 545 ppm to 7 ppm) when compared to the material loaded onto the hydroxyapatite column.
  • the column was stripped using three column volumes of 0.3 molar sodium phosphate, pH 6.8, rinsed with a half column volume of 25 millimolar sodium phosphate, pH 6.8, regenerated using two column volumes of 1 molar sodium hydroxide, rinsed with three column volumes of 0.1 molar sodium hydroxide, 10 millimolar sodium phosphate, and stored in the same solution, in which condition it is ready for reuse.
  • Figure 1 shows the absorbance and conductivity profiles of the column.
  • Most of the protein in the sample flows through the column before the increase in conductivity (to about 20 mS) caused by the addition 0.3 molar sodium phosphate, between about 55 and 59 column volumes.
  • the large increase in conductance (to about 200 mS) thereafter corresponds to the addition of 1.0 molar sodium hydroxide to regenerate the column as explained above.
  • TNFR:F C constitutes the vast majority species in the protein sample loaded onto the column, this profile indicates that TNFR:F C flows through a hydroxyapatite column run in 25 millimolar sodium phosphate, pH 6.8.
  • a column of ceramic hydroxyapatite (Type II, Bio-Rad, 80 ⁇ ) 18 cm in height and 1.6 cm in internal diameter was pre-equilibrated with two column volumes of 0.4 M sodium phosphate, pH 6.8 and equilibrated with three column volumes of 25 millimolar sodium phosphate, pH 6.8.
  • the amounts of Protein A and PRI in the sample were determined using ELISA assays.
  • the flow-through liquid, containing TNFR:F C was collected.
  • the column was washed with three column volumes of 25 millimolar sodium phosphate, pH 6.8, and the wash was collected. Almost all (99-100%) of the TNFR:F C loaded was recovered in the flow-through liquid plus the wash.
  • the amount of Protein A and PRI in the sample was reduced by 5.3-5.4 fold and 2.8-3.7 fold, respectively, when compared to the material loaded onto the hydroxyapatite column.
  • the column was stripped using five column volumes of 0.4 molar sodium phosphate, pH 6.8, regenerated using two column volumes of 1 M sodium hydroxide, rinsed in three column volumes of 10 millimolar sodium phosphate, pH 6.8, 0.1 M sodium hydroxide, and stored in the same solution, in which condition it is ready for reuse.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Medicinal Chemistry (AREA)
  • Zoology (AREA)
  • Immunology (AREA)
  • Toxicology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Cell Biology (AREA)
  • Analytical Chemistry (AREA)
  • Peptides Or Proteins (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Claims (21)

  1. Verfahren für die Reinigung eines Proteins aus einer Probe umfassend das Protein und mindestens eine Proteinverunreinigung, welches umfasst, dass die Probe einer Hydroxyapatit-Chromatographie unterzogen wird, wobei das Protein von mindestens einer Proteinverunreinigung getrennt wird und wobei die Mehrzahl der Proteinmoleküle im Durchfluss und in der Waschflüssigkeit wiedergefunden wird, wobei das Protein zuvor mit Hilfe von Affinitätschromatographie unter Verwendung eines zweiten Proteins, welches an einen festen Träger als Adsorbent gebunden ist, gereinigt wird, wobei das zweite Protein spezifisch an eine konstante Immunglobulindomäne eines Antikörpers bindet und wobei das Protein eine konstante Immunglobulindomäne eines Antikörpers umfasst.
  2. Verfahren für die Reinigung eines Proteins, welches umfasst:
    (a) Das Protein und mindestens eine Proteinverunreinigung werden einer Affinitätschromatographie unterzogen unter Verwendung eines zweiten Proteins, welches an einen festen Träger als Adsorbent gebunden ist, wobei das zweite Protein spezifisch an eine konstante Immunglobulindomäne eines Antikörpers bindet und wobei das Protein eine konstante Immunglobulindomäne eines Antikörpers umfasst, an welche sich das zweite Protein spezifisch bindet, und
    (b) das Protein und mindestens eine Proteinverunreinigung werden anschließend einer Hydroxyapatit-Chromatographie unterzogen, wobei die Mehrzahl der Proteinmoleküle im Durchfluss und in der Waschflüssigkeit wiedergefunden wird, wobei das Protein mindestens von einer Proteinverunreinigung getrennt wird.
  3. Verfahren nach Anspruch 1 oder 2, wobei das zweite Protein in nachweisbarer Menge in der Probe vorhanden ist und wobei das Protein mit Hilfe von Hydroxyapatit-Chromatographie von dem zweiten Protein getrennt wird.
  4. Verfahren nach Anspruch 1, 2 oder 3, wobei das Protein durch kultivierte Tierzellen in ein Kulturmedium sezerniert wird.
  5. Verfahren nach Anspruch 4, wobei es sich bei den Tierzellen um CHO-Zellen handelt.
  6. Verfahren nach einem der Ansprüche 2 bis 5, wobei das Protein einen Fc-Teil eines Antikörpers umfasst.
  7. Verfahren nach Anspruch 6, wobei es sich bei dem Protein um TNFR:Fc oder einen Antikörper handelt.
  8. Verfahren nach Anspruch 1 oder 2, wobei das zweite Protein aus der Gruppe ausgewählt ist, die aus Protein A und Protein G besteht.
  9. Verfahren nach einem der Ansprüche 1 bis 8, wobei die Hydroxyapatit-Chromatographie in einer Lösung durchgeführt wird, welche Natriumphosphat in einer Konzentration zwischen etwa 5 millimolar und etwa 50 millimolar enthält und einen pH-Wert zwischen etwa 6,0 und etwa 8,6 aufweist.
  10. Verfahren für die Reinigung eines rekombinanten Fusionsproteins aus einer Probe enthaltend das rekombinante Fusionsprotein und mindestens eine Proteinverunreinigung, welches umfasst, dass die Probe einer Hydroxyapatit-Chromatographie unterzogen wird, wobei das rekombinante Fusionsprotein einen Fc-Teil eines Antikörpers und ein Nicht-Antikörper-Protein umfasst, wobei das rekombinante Fusionsprotein zuvor mit Hilfe einer Affinitätschromatographie unter Verwendung eines zweiten Proteins, welches an einen festen Träger als Adsorbent gebunden ist, gereinigt wird, und wobei die Mehrzahl der Moleküle des rekombinanten Fusionsproteins im Durchfluss und in der Waschflüssigkeit wiedergefunden wird.
  11. Verfahren für die Reinigung eines rekombinanten Fusionsproteins, welches umfasst, dass das rekombinante Fusionsprotein und mindestens eine Proteinverunreinigung einer Affinitätschromatographie unterzogen werden unter Verwendung eines zweiten Proteins, welches an einen festen Träger als Adsorbent gebunden wird, bevor das rekombinante Fusionsprotein und mindestens eine Proteinverunreinigung einer Hydroxyapatit-Chromatographie unterzogen werden, wobei die Mehrzahl der Moleküle des rekombinanten Fusionsproteins im Durchfluss und in der Waschflüssigkeit wiedergefunden werden und wobei das rekombinante Fusionsprotein einen Fc-Teil eines Antikörpers und ein Nicht-Antikörper-Protein umfasst.
  12. Verfahren nach Anspruch 10 oder 11, wobei die Mehrzahl der Moleküle des rekombinanten Fusionsproteins nach der Hydroxyapatit-Chromatographie im Durchfluss und in der Waschflüssigkeit wiedergefunden wird.
  13. Verfahren nach einem der Ansprüche 10 bis 12, wobei es sich bei dem rekombinanten Fusionsprotein um TNFR:Fc handelt.
  14. Verfahren nach einem der Ansprüche 10 bis 13, wobei das zweite Protein aus der Gruppe ausgewählt ist, welche aus Protein A und Protein G besteht.
  15. Verfahren nach einem der Ansprüche 10 bis 14, wobei die Hydroxyapatit-Chromatographie in einer Lösung durchgeführt wird, welche Natriumphosphat in einer Konzentration zwischen etwa 5 millimolar und etwa 50 millimolar enthält und einen pH-Wert zwischen etwa 6,0 und etwa 8,6 aufweist.
  16. Verfahren nach einem der Ansprüche 10 bis 15, wobei das rekombinante Fusionsprotein durch kultivierte Tierzellen in ein Kulturmedium sezerniert wird.
  17. Verfahren nach Anspruch 16, wobei es sich bei den Tierzellen um CHO-Zellen handelt.
  18. Verfahren für die Reinigung von TNFR:Fc, welches umfasst, dass eine Probe enthaltend TNFR:Fc und mindestens eine Proteinverunreinigung einer Hydroxyapatit-Chromatographie unterzogen wird unter Bedingungen, bei welchen die Mehrzahl der Moleküle von TNFR:Fc im Durchfluss und in der Waschflüssigkeit wiedergefunden wird, wobei mindestens eine Proteinverunreinigung von TNFR:FC getrennt wird.
  19. Verfahren für die Trennung eines Proteins von Proteinen einer Wirtszelle, welches umfasst, dass eine Probe enthaltend das Protein und mindestens ein Protein einer Wirtszelle einer Hydroxyapatit-Chromatographie unterzogen wird, wobei mindestens ein Protein einer Wirtszelle von dem Protein getrennt wird, wobei das Protein eine konstante Immunglobulindomäne eines Antikörpers umfasst, wobei die Mehrzahl der Moleküle des Proteins im Durchfluss und in der Waschflüssigkeit wiedergefunden wird und wobei sowohl das Protein als auch das Protein der Wirtszelle durch kultivierte Tierzellen in ein Kulturmedium sezerniert werden.
  20. Verfahren nach Anspruch 19, wobei die Konzentration der Proteine der Wirtszelle bezogen auf das Protein im Durchfluss und in der Waschflüssigkeit weniger als etwa 100 Teile pro Million beträgt.
  21. Verfahren nach Anspruch 19 oder 20, wobei es sich bei dem Protein um TNFR:Fc handelt.
EP02794380A 2001-12-21 2002-12-20 Proteinreinigungsverfahren Expired - Lifetime EP1463942B2 (de)

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
US34336301P 2001-12-21 2001-12-21
US343363P 2001-12-21
US34718902P 2002-01-08 2002-01-08
US347189P 2002-01-08
US36427202P 2002-03-12 2002-03-12
US364272P 2002-03-12
PCT/US2002/041159 WO2003059935A2 (en) 2001-12-21 2002-12-20 Methods for purifying protein

Publications (4)

Publication Number Publication Date
EP1463942A2 EP1463942A2 (de) 2004-10-06
EP1463942A4 EP1463942A4 (de) 2005-01-19
EP1463942B1 true EP1463942B1 (de) 2009-03-18
EP1463942B2 EP1463942B2 (de) 2012-06-20

Family

ID=27407561

Family Applications (1)

Application Number Title Priority Date Filing Date
EP02794380A Expired - Lifetime EP1463942B2 (de) 2001-12-21 2002-12-20 Proteinreinigungsverfahren

Country Status (11)

Country Link
US (2) US7122641B2 (de)
EP (1) EP1463942B2 (de)
JP (1) JP4363986B2 (de)
AT (1) ATE425989T1 (de)
AU (1) AU2002359816B2 (de)
CA (1) CA2469815C (de)
DE (1) DE60231651D1 (de)
ES (1) ES2322945T5 (de)
MX (1) MXPA04006053A (de)
PL (1) PL374311A1 (de)
WO (1) WO2003059935A2 (de)

Families Citing this family (73)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7329388B2 (en) 1999-11-08 2008-02-12 Princeton Biochemicals, Inc. Electrophoresis apparatus having staggered passage configuration
US6406604B1 (en) 1999-11-08 2002-06-18 Norberto A. Guzman Multi-dimensional electrophoresis apparatus
MXPA04006053A (es) * 2001-12-21 2005-03-31 Immunex Corp Metodos para purificacion de proteina.
SI2261230T1 (sl) 2002-09-11 2017-08-31 Chugai Seiyaku Kabushiki Kaisha Postopek čiščenja proteinov
GB0304576D0 (en) * 2003-02-28 2003-04-02 Lonza Biologics Plc Protein a chromatography
US7850970B2 (en) 2003-08-26 2010-12-14 The Regents Of The University Of Colorado Inhibitors of serine protease activity and their use in methods and compositions for treatment of bacterial infections
CN1898266B (zh) * 2003-10-27 2011-09-21 惠氏公司 使用羟磷灰石层析除去高分子量聚集体
WO2005047882A2 (en) 2003-11-07 2005-05-26 Princeton Biochemicals, Inc. Multi-dimensional electrophoresis apparatus
US8007725B2 (en) 2003-11-07 2011-08-30 Princeton Biochemicals, Inc. Electrophoresis apparatus having valve system
WO2005071410A1 (en) * 2004-01-13 2005-08-04 Tanox, Inc. Detecting and quantifying host cell proteins in recombinant protein products
JP4872327B2 (ja) * 2004-11-26 2012-02-08 東レ株式会社 改質されたインターロイキン−11とその製造方法
CN104610422A (zh) 2005-03-11 2015-05-13 惠氏公司 弱分配层析的方法
CN101198862A (zh) * 2005-04-20 2008-06-11 谢佳辉 Ph梯度离子交换lc-ms和与质谱相容的缓冲液
EP1909831A4 (de) 2005-06-14 2013-02-20 Amgen Inc Selbstpuffernde proteinformulierungen
US7375188B2 (en) 2005-07-29 2008-05-20 Mallinckrodt Baker, Inc. Vegetarian protein a preparation and methods thereof
MY185597A (en) * 2005-09-20 2021-05-24 Avantor Performance Mat Inc Vegetarian protein a preparation and methods thereof
EP1816201A1 (de) 2006-02-06 2007-08-08 CSL Behring GmbH Modifizierter Koagulationsfaktor VIIa mit verbesserter 'half-life'-Stabiltät
EP2004689A4 (de) 2006-04-05 2010-06-02 Abbott Biotech Ltd Antikörperreinigung
WO2007144173A1 (en) 2006-06-14 2007-12-21 Csl Behring Gmbh Proteolytically cleavable fusion protein comprising a blood coagulation factor
US7939632B2 (en) 2006-06-14 2011-05-10 Csl Behring Gmbh Proteolytically cleavable fusion proteins with high molar specific activity
JP2010510963A (ja) * 2006-06-14 2010-04-08 グラクソスミスクライン・リミテッド・ライアビリティ・カンパニー セラミックヒドロキシアパタイトを使用する抗体の精製方法
EP1867660A1 (de) 2006-06-14 2007-12-19 CSL Behring GmbH Fusionsprotein welches proteolytisch geschnitten werden kann und einen Faktor der Blutgerinnungskaskade enthält
ZA200900836B (en) * 2006-08-28 2010-05-26 Ares Trading Sa Process for the purification of FC-fusion proteins
ES2755386T5 (es) 2006-08-28 2023-04-05 Ares Trading Sa Proceso para la purificación de proteínas que contienen fragmentos Fc
JP2010501622A (ja) * 2006-08-28 2010-01-21 アレス トレーディング ソシエテ アノニム Fc−融合タンパク質の精製法
WO2008033517A2 (en) 2006-09-13 2008-03-20 Abbott Laboratories Cell culture improvements
US8911964B2 (en) 2006-09-13 2014-12-16 Abbvie Inc. Fed-batch method of making human anti-TNF-alpha antibody
US20100047252A1 (en) * 2006-11-22 2010-02-25 3M Innovative Properties Company Antibody with protein a selectivity
US7488807B2 (en) * 2006-11-22 2009-02-10 3M Innovative Properties Company Antibody with protein A selectivity
JP5448839B2 (ja) 2006-12-22 2014-03-19 ツェー・エス・エル・ベーリング・ゲー・エム・ベー・ハー インビボで長い半減期を有する修飾された凝固因子
EP2238154B1 (de) 2008-01-18 2015-09-16 Bio-Rad Laboratories, Inc. Erweiterte reinigung phosphorylierter und nicht phosphorylierter biomoleküle durch apatit-chromatographie
US20100058173A1 (en) * 2008-08-28 2010-03-04 Kabushiki Kaisha Toshiba Display processing apparatus, display processing method, and computer program product
JP5632376B2 (ja) * 2008-09-23 2014-11-26 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft エリスロポエチンの精製
EP2921501A1 (de) * 2008-10-20 2015-09-23 Abbvie Inc. Isolation und Aufreinigung von Antikörpern mit Protein-A-Affinitätschromatografie
CN104974251A (zh) 2008-10-20 2015-10-14 Abbvie公司 在抗体纯化过程中的病毒灭活
ES2699434T3 (es) 2008-10-31 2019-02-11 Wyeth Llc Purificación de proteínas ácidas utilizando cromatografía de hidroxiapatita cerámica
US8945895B2 (en) 2009-07-31 2015-02-03 Baxter International Inc. Methods of purifying recombinant ADAMTS13 and other proteins and compositions thereof
US8722615B2 (en) * 2009-12-02 2014-05-13 Acceleron Pharma, Inc. Compositions and methods for increasing serum half-life
CA2787009C (en) 2010-01-15 2018-04-03 Bio-Rad Laboratories, Inc. Surface neutralization of apatite
BR112012030530A2 (pt) * 2010-05-18 2022-12-20 Abbvie Inc Aparelho e processo para purificação de proteínas
US8895707B2 (en) 2010-08-18 2014-11-25 Bio-Rad Laboratories, Inc. Elution of proteins from hydroxyapatite resins without resin deterioration
US9592540B2 (en) 2011-02-02 2017-03-14 Bio-Rad Laboratories, Inc. Apatite surface neutralization with alkali solutions
EP2702077A2 (de) 2011-04-27 2014-03-05 AbbVie Inc. Verfahren zur steuerung des galactosylierungsprofil von rekombinant exprimierten proteinen
US8911992B2 (en) * 2011-04-28 2014-12-16 Bio-Rad Laboratories, Inc. DNA removal in target molecule purification
WO2012170938A1 (en) 2011-06-08 2012-12-13 Acceleron Pharma Inc. Compositions and methods for increasing serum half-life
WO2012178102A2 (en) 2011-06-24 2012-12-27 The Regents Of The Unversity Of Colorado, A Body Corporate Compositions, methods and uses for alpha-1 antitrypsin fusion molecules
KR20140043934A (ko) * 2011-07-08 2014-04-11 머크 샤프 앤드 돔 코포레이션 Fc-융합 단백질의 정제 방법
JP2015504675A (ja) 2012-01-10 2015-02-16 ザ リージェンツ オブ ザ ユニバーシティ オブ コロラド,ア ボディー コーポレイトTHE REGENTS OF THE UNIVERSITY OF COLORADO,a body corporate アルファ−1アンチトリプシン融合分子の組成物、方法、及び使用
WO2013158279A1 (en) 2012-04-20 2013-10-24 Abbvie Inc. Protein purification methods to reduce acidic species
WO2013158273A1 (en) 2012-04-20 2013-10-24 Abbvie Inc. Methods to modulate c-terminal lysine variant distribution
US9067990B2 (en) 2013-03-14 2015-06-30 Abbvie, Inc. Protein purification using displacement chromatography
WO2013176754A1 (en) 2012-05-24 2013-11-28 Abbvie Inc. Novel purification of antibodies using hydrophobic interaction chromatography
CN104507952B (zh) 2012-05-30 2018-08-10 生物辐射实验室股份有限公司 基于磷灰石的色谱树脂的原位恢复
WO2014035475A1 (en) 2012-09-02 2014-03-06 Abbvie Inc. Methods to control protein heterogeneity
US9512214B2 (en) 2012-09-02 2016-12-06 Abbvie, Inc. Methods to control protein heterogeneity
BR112015005389B1 (pt) 2012-09-13 2022-10-18 Indiana University Research And Technology Corporation Molécula de ácido nucleico recombinante, proteína de substrato modificada de uma protease específica de patógeno de planta expressa através do patógeno de planta, vetor, e método para proteger uma planta da infecção por um patógeno de planta que secreta pelo menos uma protease específica
AU2013381687A1 (en) 2013-03-12 2015-09-24 Abbvie Inc. Human antibodies that bind human TNF-alpha and methods of preparing the same
US10023608B1 (en) 2013-03-13 2018-07-17 Amgen Inc. Protein purification methods to remove impurities
US9017687B1 (en) 2013-10-18 2015-04-28 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same using displacement chromatography
WO2014159579A1 (en) 2013-03-14 2014-10-02 Abbvie Inc. MUTATED ANTI-TNFα ANTIBODIES AND METHODS OF THEIR USE
WO2014159441A1 (en) * 2013-03-14 2014-10-02 Amgen Inc. Removal of leaked affinity purification ligand
US9499614B2 (en) 2013-03-14 2016-11-22 Abbvie Inc. Methods for modulating protein glycosylation profiles of recombinant protein therapeutics using monosaccharides and oligosaccharides
US9598667B2 (en) 2013-10-04 2017-03-21 Abbvie Inc. Use of metal ions for modulation of protein glycosylation profiles of recombinant proteins
US9181337B2 (en) 2013-10-18 2015-11-10 Abbvie, Inc. Modulated lysine variant species compositions and methods for producing and using the same
US9085618B2 (en) 2013-10-18 2015-07-21 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US8946395B1 (en) 2013-10-18 2015-02-03 Abbvie Inc. Purification of proteins using hydrophobic interaction chromatography
US20150139988A1 (en) 2013-11-15 2015-05-21 Abbvie, Inc. Glycoengineered binding protein compositions
EP3157689B1 (de) 2014-06-23 2021-04-28 Bio-Rad Laboratories, Inc. Apatit-vorbehandlung
EP3157551B1 (de) 2014-06-23 2023-02-15 Bio-Rad Laboratories, Inc. In-situ-restaurierung von apatit
GB2528301A (en) * 2014-07-17 2016-01-20 Univ Newcastle Biomolecule separation using apatite
WO2018063980A1 (en) * 2016-09-29 2018-04-05 Bio-Rad Laboratories, Inc. Agarose-filled ceramic apatite
US20180110856A1 (en) 2016-10-21 2018-04-26 Amgen Inc. Pharmaceutical Formulations and Methods of Making the Same
WO2022140389A1 (en) 2020-12-22 2022-06-30 Amgen Inc. Cell culture method

Family Cites Families (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4745183A (en) * 1984-02-21 1988-05-17 Bio-Rad Laboratories, Inc. Use of hydroxylapatite for the analysis and preparation of purified monoclonal antibodies
DE3738541A1 (de) * 1987-11-13 1989-05-24 Hoechst Ag Verfahren zur isolierung und reinigung von hirudin
JPH0774231B2 (ja) 1986-12-11 1995-08-09 東燃株式会社 抗体の分離方法
US4933286A (en) * 1987-03-30 1990-06-12 Plant Cell Research Institute, Inc. Purification of apase-11 and retrieval of the nematode resistance gene
US5115101A (en) * 1988-06-08 1992-05-19 Miles Inc. Removal of protein A from antibody preparations
JPH022937A (ja) 1988-06-17 1990-01-08 Toray Ind Inc 酵素標識抗体の分離精製方法
JPH03139292A (ja) 1989-05-22 1991-06-13 Toray Ind Inc ヒトインタ―ロイキン―6に対するモノクロ―ナル抗体
US5395760A (en) * 1989-09-05 1995-03-07 Immunex Corporation DNA encoding tumor necrosis factor-α and -β receptors
DK0417563T3 (da) * 1989-09-12 2000-11-06 Hoffmann La Roche TNF-bindende proteiner
JPH03291300A (ja) 1989-12-28 1991-12-20 Mitsui Toatsu Chem Inc ポリクローナル抗体の分離方法
GB9007657D0 (en) * 1990-04-04 1990-05-30 Connaught Lab Purification of a pertussis outer membrane protein(omp69)
JPH04368399A (ja) 1991-06-12 1992-12-21 Toagosei Chem Ind Co Ltd 抗体の分離方法
JP2961986B2 (ja) 1991-08-22 1999-10-12 東亞合成株式会社 ポリクローナル抗体の分離精製方法
DE4205938A1 (de) * 1992-02-27 1993-09-02 Medac Klinische Spezialpraep Immunotoxine
JP3178080B2 (ja) 1992-04-28 2001-06-18 チッソ株式会社 抗体の分離精製方法
US5278284A (en) * 1992-05-14 1994-01-11 Miller Brewing Company Protein purification method
US6099830A (en) * 1994-08-09 2000-08-08 Zymogenetics, Inc. Methods for stimulating erythropoiesis using hematopoietic proteins
US5641655A (en) * 1994-11-30 1997-06-24 Zymogenetics, Inc. Methods for producing thrombopoietin polypeptides using a mammalian tissue plasminogen activator secretory peptide
JPH08188599A (ja) 1995-01-09 1996-07-23 Toagosei Co Ltd IgY抗体の分離精製方法
US5744587A (en) * 1995-06-07 1998-04-28 Zymogenetics, Inc. Method for purifying thrombopoietin
PT950067E (pt) * 1996-11-27 2007-12-06 Genentech Inc Purificação por afinidade de um polipéptido numa matriz de proteína a.
EP0998486B2 (de) * 1997-06-13 2012-02-01 Genentech, Inc. Proteinreinigung mittels chromatographie und darauffolgende filtration auf beladener schicht
MXPA04006053A (es) * 2001-12-21 2005-03-31 Immunex Corp Metodos para purificacion de proteina.
WO2003059936A2 (en) * 2002-01-10 2003-07-24 Syngenta Participations Ag Phytophthora Infestans CDC 14 Phosphatase Gene, Protein and Methods of Use

Also Published As

Publication number Publication date
WO2003059935A2 (en) 2003-07-24
AU2002359816B2 (en) 2006-07-13
CA2469815A1 (en) 2003-07-24
CA2469815C (en) 2011-05-03
PL374311A1 (en) 2005-10-03
ES2322945T5 (es) 2012-10-17
JP4363986B2 (ja) 2009-11-11
DE60231651D1 (de) 2009-04-30
WO2003059935A3 (en) 2003-10-16
AU2002359816A1 (en) 2003-07-30
ATE425989T1 (de) 2009-04-15
ES2322945T3 (es) 2009-07-02
MXPA04006053A (es) 2005-03-31
US20070010661A1 (en) 2007-01-11
EP1463942A4 (de) 2005-01-19
JP2005538686A (ja) 2005-12-22
US7122641B2 (en) 2006-10-17
EP1463942A2 (de) 2004-10-06
EP1463942B2 (de) 2012-06-20
US20030166869A1 (en) 2003-09-04
US7476722B2 (en) 2009-01-13

Similar Documents

Publication Publication Date Title
EP1463942B1 (de) Proteinreinigungsverfahren
EP1687328B1 (de) Verfahren zur aufreinigung von proteinen in einer durchflussfraktion aus der chromatographie mit hydrophoben wechselwirkungen
US11492372B2 (en) Removal of leaked affinity purification ligand
EP2726496B1 (de) Verfahren zum trennen von monomeren polypeptiden von aggregierten polypeptiden
US8273707B2 (en) Process for purifying proteins

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20040615

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR IE IT LI LU MC NL PT SE SI SK TR

AX Request for extension of the european patent

Extension state: AL LT LV MK RO

A4 Supplementary search report drawn up and despatched

Effective date: 20041207

RIC1 Information provided on ipc code assigned before grant

Ipc: 7C 07K 1/22 B

Ipc: 7C 07K 14/705 A

17Q First examination report despatched

Effective date: 20050307

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR IE IT LI LU MC NL PT SE SI SK TR

AX Request for extension of the european patent

Extension state: AL LT LV MK RO

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REF Corresponds to:

Ref document number: 60231651

Country of ref document: DE

Date of ref document: 20090430

Kind code of ref document: P

REG Reference to a national code

Ref country code: CH

Ref legal event code: NV

Representative=s name: SCHMAUDER & PARTNER AG PATENTANWALTSBUERO

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2322945

Country of ref document: ES

Kind code of ref document: T3

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20090318

Ref country code: NL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20090318

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20090318

REG Reference to a national code

Ref country code: CH

Ref legal event code: PCAR

Free format text: SCHMAUDER & PARTNER AG PATENT- UND MARKENANWAELTE VSP;ZWAENGIWEG 7;8038 ZUERICH (CH)

LTIE Lt: invalidation of european patent or patent extension

Effective date: 20090318

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20090318

Ref country code: SE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20090618

NLV1 Nl: lapsed or annulled due to failure to fulfill the requirements of art. 29p and 29m of the patents act
PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20090318

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20090826

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20090318

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20090318

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20090318

PLBI Opposition filed

Free format text: ORIGINAL CODE: 0009260

PLAX Notice of opposition and request to file observation + time limit sent

Free format text: ORIGINAL CODE: EPIDOSNOBS2

26 Opposition filed

Opponent name: SANDOZ AG (CH)/ SANDOZ GMBH (AT)

Effective date: 20091218

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20090318

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20090618

PLAF Information modified related to communication of a notice of opposition and request to file observations + time limit

Free format text: ORIGINAL CODE: EPIDOSCOBS2

PLBB Reply of patent proprietor to notice(s) of opposition received

Free format text: ORIGINAL CODE: EPIDOSNOBS3

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20100701

PLAB Opposition data, opponent's data or that of the opponent's representative modified

Free format text: ORIGINAL CODE: 0009299OPPO

R26 Opposition filed (corrected)

Opponent name: SANDOZ AG (CH)/ SANDOZ GMBH (AT)

Effective date: 20091218

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20090619

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20091220

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20090318

PLAB Opposition data, opponent's data or that of the opponent's representative modified

Free format text: ORIGINAL CODE: 0009299OPPO

PLAB Opposition data, opponent's data or that of the opponent's representative modified

Free format text: ORIGINAL CODE: 0009299OPPO

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20090318

R26 Opposition filed (corrected)

Opponent name: SANDOZ AG (CH)/ SANDOZ GMBH (AT)

Effective date: 20091218

R26 Opposition filed (corrected)

Opponent name: SANDOZ AG (CH)/ SANDOZ GMBH (AT)

Effective date: 20091218

PUAH Patent maintained in amended form

Free format text: ORIGINAL CODE: 0009272

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: PATENT MAINTAINED AS AMENDED

27A Patent maintained in amended form

Effective date: 20120620

AK Designated contracting states

Kind code of ref document: B2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR IE IT LI LU MC NL PT SE SI SK TR

AX Request for extension of the european patent

Extension state: AL LT LV MK RO

REG Reference to a national code

Ref country code: CH

Ref legal event code: AEN

Free format text: AUFRECHTERHALTUNG DES PATENTES IN GEAENDERTER FORM

REG Reference to a national code

Ref country code: DE

Ref legal event code: R102

Ref document number: 60231651

Country of ref document: DE

Effective date: 20120620

REG Reference to a national code

Ref country code: ES

Ref legal event code: DC2A

Ref document number: 2322945

Country of ref document: ES

Kind code of ref document: T5

Effective date: 20121017

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 14

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 15

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 16

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: IT

Payment date: 20201110

Year of fee payment: 19

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: DE

Payment date: 20211102

Year of fee payment: 20

Ref country code: FR

Payment date: 20211115

Year of fee payment: 20

Ref country code: IE

Payment date: 20211109

Year of fee payment: 20

Ref country code: GB

Payment date: 20211104

Year of fee payment: 20

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: CH

Payment date: 20211116

Year of fee payment: 20

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: ES

Payment date: 20220105

Year of fee payment: 20

REG Reference to a national code

Ref country code: DE

Ref legal event code: R071

Ref document number: 60231651

Country of ref document: DE

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

REG Reference to a national code

Ref country code: GB

Ref legal event code: PE20

Expiry date: 20221219

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: GB

Free format text: LAPSE BECAUSE OF EXPIRATION OF PROTECTION

Effective date: 20221219

REG Reference to a national code

Ref country code: IE

Ref legal event code: MK9A

REG Reference to a national code

Ref country code: ES

Ref legal event code: FD2A

Effective date: 20230426

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IE

Free format text: LAPSE BECAUSE OF EXPIRATION OF PROTECTION

Effective date: 20221220

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20211231

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: ES

Free format text: LAPSE BECAUSE OF EXPIRATION OF PROTECTION

Effective date: 20221221